Blocking P2X7 by intracerebroventricular injection of P2X7-specific nanobodies reduces stroke lesions

J Neuroinflammation. 2022 Oct 12;19(1):256. doi: 10.1186/s12974-022-02601-z.

Abstract

Background: Previous studies have demonstrated that purinergic receptors could be therapeutic targets to modulate the inflammatory response in multiple models of brain diseases. However, tools for the selective and efficient targeting of these receptors are lacking. The development of new P2X7-specific nanobodies (nbs) has enabled us to effectively block the P2X7 channel.

Methods: Temporary middle cerebral artery occlusion (tMCAO) in wild-type (wt) and P2X7 transgenic (tg) mice was used to model ischemic stroke. Adenosine triphosphate (ATP) release was assessed in transgenic ATP sensor mice. Stroke size was measured after P2X7-specific nbs were injected intravenously (iv) and intracerebroventricularly (icv) directly before tMCAO surgery. In vitro cultured microglia were used to investigate calcium influx, pore formation via 4,6-diamidino-2-phenylindole (DAPI) uptake, caspase 1 activation and interleukin (IL)-1β release after incubation with the P2X7-specific nbs.

Results: Transgenic ATP sensor mice showed an increase in ATP release in the ischemic hemisphere compared to the contralateral hemisphere or the sham-treated mice up to 24 h after stroke. P2X7-overexpressing mice had a significantly greater stroke size 24 h after tMCAO surgery. In vitro experiments with primary microglial cells demonstrated that P2X7-specific nbs could inhibit ATP-triggered calcium influx and the formation of membrane pores, as measured by Fluo4 fluorescence or DAPI uptake. In microglia, we found lower caspase 1 activity and subsequently lower IL-1β release after P2X7-specific nb treatment. The intravenous injection of P2X7-specific nbs compared to isotype controls before tMCAO surgery did not result in a smaller stroke size. As demonstrated by fluorescence-activated cell sorting (FACS), after stroke, iv injected nbs bound to brain-infiltrated macrophages but not to brain resident microglia, indicating insufficient crossing of the blood-brain barrier of the nbs. Therefore, we directly icv injected the P2X7-specific nbs or the isotype nbs. After icv injection of 30 µg of P2X7 specific nbs, P2X7 specific nbs bound sufficiently to microglia and reduced stroke size.

Conclusion: Mechanistically, we can show that there is a substantial increase of ATP locally after stroke and that blockage of the ATP receptor P2X7 by icv injected P2X7-specific nbs can reduce ischemic tissue damage.

Keywords: Cerebrovascular disease; Ischemic stroke; Nanobodies; Nanomedicine; P2X7.

MeSH terms

  • Adenosine Triphosphate / pharmacology
  • Animals
  • Calcium / metabolism
  • Caspase 1 / metabolism
  • Infarction, Middle Cerebral Artery / pathology
  • Interleukin-1beta / metabolism
  • Mice
  • Microglia / metabolism
  • Receptors, Purinergic P2* / metabolism
  • Receptors, Purinergic P2X7 / metabolism
  • Single-Domain Antibodies* / metabolism
  • Stroke* / metabolism

Substances

  • Interleukin-1beta
  • Receptors, Purinergic P2
  • Receptors, Purinergic P2X7
  • Single-Domain Antibodies
  • Adenosine Triphosphate
  • Caspase 1
  • Calcium